UBE2D4 Human

Ubiquitin Conjugating Enzyme E2D4 Human Recombinant
Shipped with Ice Packs
In Stock

Description

UBE2D4 Human Recombinant produced in E.coli is a single, non-glycosylated polypeptide chain containing 167 amino acids (1-147) and having a molecular mass of 18.8 kDa.
UBE2D4 is fused to a 20 amino acid His-tag at N-terminus & purified by proprietary chromatographic techniques.

Product Specs

Introduction
UBE2D4, a member of the ubiquitin-conjugating enzyme family, plays a crucial role in the ubiquitination pathway. This pathway targets proteins for degradation via the proteasome. As a ubiquitin-conjugating enzyme (E2 enzyme), UBE2D4 facilitates the transfer of ubiquitin from the E1 complex to target proteins. Notably, UBE2D4 exhibits versatility in promoting polyubiquitination by utilizing all seven lysine residues on ubiquitin. However, it preferentially catalyzes the formation of 'Lys-11' and 'Lys-48'-linked polyubiquitin chains.
Description
Recombinantly produced in E.coli, UBE2D4 Human Recombinant is a single, non-glycosylated polypeptide chain. It comprises 167 amino acids (residues 1-147) and exhibits a molecular mass of 18.8 kDa. The protein features a 20 amino acid His-tag fused at the N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
A colorless solution that has undergone sterile filtration.
Formulation
The SRGN solution is provided at a concentration of 1mg/ml and contains 20mM Tris-HCl buffer (pH 8.0), 2mM DTT, 0.1M NaCl, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product should be kept at 4°C. For extended storage, freezing at -20°C is recommended. The addition of a carrier protein (0.1% HSA or BSA) is advisable for long-term storage. Repeated freeze-thaw cycles should be avoided.
Purity
The purity of the product is determined to be greater than 95% based on SDS-PAGE analysis.
Synonyms
Ubiquitin-conjugating enzyme E2D 4 (putative), UBCH5D, HBUCE1, Ubiquitin carrier protein D4, Ubiquitin-protein ligase D4, EC 6.3.2.19.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MALKRIQKEL TDLQRDPPAQ CSAGPVGDDL FHWQATIMGP NDSPYQGGVF FLTIHFPTDY PFKPPKVAFT TKIYHPNINS NGSICLDILR SQWSPALTVS KVLLSICSLL CDPNPDDPLV PEIAHTYKAD REKYNRLARE WTQKYAM

Q&A

What is the role of UBE2D4 in the ubiquitination pathway and protein homeostasis?

Recent research has demonstrated that knockdown of UBE2D/eff (the Drosophila homolog) reduces proteasomal activity in skeletal muscle, which can be rescued by expression of human UBE2D2, indicating a conserved role in maintaining proteasome function . This finding suggests that UBE2D family members, including UBE2D4, are necessary for maintaining proper protein quality control systems during aging and in disease contexts. The capacity of UBE2D enzymes to modulate proteasome activity likely explains their role in proteostasis during skeletal muscle aging as well as in the context of proteinopathies like Huntington's disease .

Methodologically, researchers investigating UBE2D4's role in proteostasis should consider both loss-of-function approaches (RNAi, CRISPR knockout) and rescue experiments with human UBE2D4 or other family members, coupled with proteasome activity assays and proteomics analysis.

How conserved is UBE2D4 across species, and what experimental approaches can demonstrate functional conservation?

UBE2D4 demonstrates significant evolutionary conservation, with clear homologs in model organisms including Drosophila. The Drosophila homolog "eff" shows a DIOPT homology score of 10 with human UBE2D4, indicating substantial sequence similarity . More importantly, functional conservation has been demonstrated through cross-species rescue experiments.

In experimental systems, human UBE2D4 can rescue retinal degeneration induced by knockdown of the Drosophila homolog eff . Specifically, when eff is knocked down in Drosophila, characteristic "rough eye" appearance (indicative of photoreceptor death), increased necrotic patches, and loss of pigmentation occur. Transgenic expression of human UBE2D4 rescues the depigmentation phenotype caused by eff knockdown, though to a lesser extent than UBE2D2 . This functional complementation approach provides strong evidence for conservation of molecular function across species.

For researchers studying conservation of UBE2D4, a combination of sequence analysis, structural modeling, and cross-species functional rescue experiments represents the most robust methodological approach. When designing such experiments, controls should include mock overexpression (e.g., mCherry) and other UBE2D family members to assess the specificity of the rescue effect.

How do researchers distinguish between UBE2D4 and other UBE2D family members in experimental settings?

Distinguishing between UBE2D family members presents significant technical challenges due to their high sequence similarity. UBE2D1, UBE2D2, UBE2D3, and UBE2D4 share extensive sequence identity, making specific detection and functional analysis difficult. For example, the tryptic peptide sequence bearing C111 (a key residue in these enzymes) is identical across all four UBE2D isoforms, complicating mass spectrometry-based identification .

Methodologically, researchers can employ several strategies to overcome this challenge:

  • Isoform-specific knockdown followed by rescue with RNAi-resistant constructs

  • Use of isoform-specific antibodies (though these may have cross-reactivity)

  • CRISPR-based tagging of endogenous UBE2D4

  • Targeted expression of UBE2D4 in knockout backgrounds

  • Analysis of tissue-specific expression patterns to identify contexts where UBE2D4 predominates

When interpreting data from proteomics experiments involving UBE2D family members, researchers should acknowledge the limitation that most analysis techniques cannot distinguish between individual UBE2D isoforms without specialized approaches. For example, in isoDTB-ABPP studies, the identical tryptic peptide sequence bearing C111 cannot differentiate between UBE2D1, UBE2D2, UBE2D3, and UBE2D4 .

What methodologies are most effective for investigating UBE2D4 substrate specificity?

Determining UBE2D4 substrate specificity requires sophisticated experimental approaches that can distinguish direct targets from secondary effects of altered proteostasis. TMT-based proteomics represents a powerful methodology for identifying proteins modulated by UBE2D activity . This approach allows for multiplexed quantitative analysis of protein changes following UBE2D knockdown versus control conditions, and can further assess whether these changes are rescued by expression of human UBE2D.

In a study using this approach with UBE2D/eff knockdown in Drosophila, several categories of proteins were identified as being significantly regulated, including proteasome components, chaperones, deubiquitinases, ubiquitin ligases, and extracellular proteins (upregulated), as well as peptidases, lipases, and secreted proteins (downregulated) . Specific proteins like Arc1, Arc2, Gnmt, and CG4594 were increased upon eff RNAi but reduced by concomitant rescue with human UBE2D2, suggesting they may be direct or indirect targets of UBE2D-mediated regulation .

For researchers investigating UBE2D4 substrate specificity, experimental designs should include:

  • RNAi-mediated knockdown of UBE2D4 with appropriate controls

  • Expression of RNAi-resistant UBE2D4 for rescue experiments

  • TMT-based or other multiplexed quantitative proteomics

  • Validation of candidate substrates through biochemical ubiquitination assays

  • Analysis of ubiquitination sites on candidate substrates

Additionally, researchers should consider UBE2D4's interactions with different E3 ligases, as these partnerships largely determine substrate specificity in the ubiquitination cascade.

How can researchers develop and validate selective inhibitors of UBE2D4 for studying its functions?

Development of selective UBE2D4 inhibitors represents a significant challenge but offers substantial research value. Recent approaches have focused on creating inhibitors that exploit multiple interaction surfaces on UBE2D enzymes. Two notable strategies include:

  • Covalent recruiters targeting UBE2D: Researchers have used covalent chemoproteomic approaches to discover compounds like EN67 that covalently modify UBE2D at specific residues, particularly C111 . These compounds can achieve partial target engagement (~24%) in cells, which may be sufficient for studying UBE2D functions given previous studies indicating sub-stoichiometric activity of heterobifunctional compounds .

  • Linked-domain protein inhibitors: This approach involves designing chimeric fusion proteins consisting of a RING/UBOX domain and a ubiquitin-like (UBL) domain that bind two sites on UBE2D simultaneously . These engineered proteins can achieve high affinity (ranging from 3×10⁻⁶M to ~1×10⁻⁹M) and function as potent inhibitors in assays of E3 ligase activity .

For validation of UBE2D inhibitors, researchers should employ multiple complementary approaches:

  • Isothermal Titration Calorimetry (ITC) to determine binding affinity

  • In vitro ubiquitination assays to assess functional inhibition

  • Yeast two-hybrid selectivity assays against a panel of E2 enzymes

  • Cellular target engagement analysis using chemoproteomics

  • Proteome-wide analysis to confirm expected functional consequences

When interpreting data from inhibitor studies, researchers should consider that most current approaches cannot distinguish between UBE2D family members due to their high sequence similarity. For example, the covalent inhibitor EN67 likely targets all four isoforms of UBE2D rather than just UBE2D4 specifically .

What role does UBE2D4 play in neurodegenerative diseases and how can it be studied experimentally?

UBE2D4 and other UBE2D family members appear to play significant roles in neurodegenerative diseases, particularly those involving protein aggregation. In Huntington's disease models, UBE2D enzymes have been shown to affect the disposal of polyglutamine-expanded huntingtin protein (Htt-polyQ) .

The experimental approach for studying UBE2D4 in neurodegenerative contexts typically involves:

  • RNAi-mediated knockdown of UBE2D in disease models

  • Assessment of protein aggregation (e.g., using fluorescently tagged polyQ proteins)

  • Rescue experiments with human UBE2D4 expression

  • Behavioral and tissue degeneration analyses in animal models

In Drosophila models, knockdown of the UBE2D homolog eff induces retinal degeneration, with characteristic "rough eye" appearance, increased necrotic patches, and pigmentation loss . This degeneration can be rescued by human UBE2D4 expression, though with somewhat lower efficacy compared to UBE2D2 .

When interpreting data from such experiments, researchers should carefully consider whether effects on protein aggregation result from direct impacts on protein disposal or from altered transgenic expression. For example, some E2 enzymes may modulate transgenic expression by regulating histone ubiquitination and degradation, which generally alters transcriptional activity . Control experiments examining mRNA levels of aggregation-prone proteins are essential to distinguish these possibilities.

How does UBE2D4 contribute to aging processes, and what methodological approaches best reveal these connections?

UBE2D enzymes, including UBE2D4, appear to play important roles in age-related proteostasis decline, particularly in tissues like skeletal muscle. Research has shown that muscle-specific knockdown of UBE2D/eff reduces organismal lifespan in Drosophila, and this effect can be partially rescued by expression of human UBE2D2 . This suggests that maintaining proper UBE2D function in muscle is necessary for optimal organismal survival.

Methodologically, researchers investigating UBE2D4's role in aging should consider:

  • Tissue-specific modulation of UBE2D4 expression (overexpression or knockdown)

  • Lifespan analyses under various conditions

  • Age-dependent assessment of proteasome activity

  • Proteomics analysis to identify age-dependent changes in UBE2D4 targets

  • Examination of UBE2D4 expression and activity across the lifespan

The proteomics approach has revealed that UBE2D/eff knockdown affects levels of specific proteins that may contribute to age-related decline in proteostasis. These include Arc1 and Arc2 (activity-regulated cytoskeleton-associated proteins), which regulate starvation-induced locomotion, neuromuscular junction function, and metabolism; Gnmt enzyme (glycine N-methyltransferase); and CG4594, an enzyme involved in fatty acid beta-oxidation .

Notably, elevation of Arc1 protein levels has been observed in Alzheimer's disease models, where it causes cytotoxicity and contributes to neuronal death . This suggests that proper regulation of Arc proteins by UBE2D enzymes may be particularly important for preventing age-related neurodegeneration.

What are the optimal experimental models for studying UBE2D4 function?

Selecting appropriate experimental models for UBE2D4 research requires careful consideration of the specific aspects being investigated. Several model systems have proven valuable for UBE2D research:

Drosophila models: The Drosophila homolog "eff" provides an excellent system for studying UBE2D function in vivo. Advantages include genetic tractability, tissue-specific knockdown capabilities, and clear phenotypic readouts (e.g., retinal degeneration, lifespan) . Cross-species rescue with human UBE2D4 allows direct assessment of functional conservation.

Cell culture systems: Human cell lines enable study of UBE2D4 in its native context. Various approaches can be employed:

  • siRNA or CRISPR-based knockdown/knockout

  • Overexpression of wild-type or mutant UBE2D4

  • Treatment with selective UBE2D inhibitors

  • Proteomics analysis following UBE2D4 modulation

In vitro biochemical systems: Purified recombinant UBE2D4 can be used in reconstituted ubiquitination assays with various E1 and E3 enzymes to assess enzymatic activity and specificity. This approach allows precise biochemical characterization but lacks cellular context.

Yeast models: Yeast two-hybrid systems have been developed to test interactions between E2s (including UBE2D4) and potential binding partners . These provide a valuable tool for screening interactions and assessing inhibitor specificity.

When designing experiments across these model systems, researchers should consider that observations in one system may not translate directly to others due to differences in expression patterns, binding partners, and regulatory mechanisms.

What proteomics approaches best characterize UBE2D4-dependent ubiquitination networks?

Several complementary proteomics approaches can effectively characterize UBE2D4-dependent ubiquitination networks:

TMT-based quantitative proteomics: This multiplexed approach allows comparison of protein levels across multiple conditions (e.g., control, UBE2D4 knockdown, rescue with human UBE2D4) . The methodology involves:

  • Sample preparation from experimental conditions

  • Tandem Mass Tag (TMT) labeling of peptides

  • Mass spectrometry analysis

  • Computational identification of significantly regulated proteins

  • Pathway enrichment analysis to identify affected biological processes

Ubiquitinome analysis: This approach specifically identifies ubiquitinated proteins and can pinpoint ubiquitination sites. Methodological considerations include:

  • Enrichment for ubiquitinated proteins (using ubiquitin antibodies or tandem ubiquitin binding entities)

  • Identification of the characteristic Gly-Gly remnant on ubiquitinated lysines

  • Quantitative comparison between control and UBE2D4-modulated conditions

Interaction proteomics: Identifying proteins that physically associate with UBE2D4 can reveal potential substrates and regulatory partners. This typically involves:

  • Immunoprecipitation of tagged UBE2D4 or antibody-based pulldown of endogenous UBE2D4

  • Mass spectrometry analysis of co-precipitated proteins

  • Validation of interactions through reciprocal pulldowns or other binding assays

Isodeuterium-tagged dibromobimane activity-based protein profiling (isoDTB-ABPP): This approach has been used to assess target engagement and selectivity of UBE2D inhibitors, which can be applied to study UBE2D4 . The technique allows identification of proteins that interact with small molecule probes and quantification of these interactions.

When interpreting proteomics data related to UBE2D4, researchers should consider that changes in protein levels may reflect direct ubiquitination targets or secondary effects resulting from altered proteostasis. Additionally, the high sequence similarity between UBE2D family members may complicate specific attribution of effects to UBE2D4 alone.

What are the methodological considerations for developing PROTACs targeting UBE2D4?

Proteolysis-targeting chimeras (PROTACs) represent an emerging approach for targeted protein degradation that could be applied to study UBE2D4 or to use UBE2D4 as a recruiting component. Several methodological considerations are important:

Using UBE2D4 as a PROTAC target:

  • Identifying selective binding ligands for UBE2D4 represents a significant challenge due to high homology with other UBE2D family members

  • Covalent recruiters like EN67 that target UBE2D enzymes provide a potential starting point for PROTAC development

  • Testing must assess degradation efficacy, specificity among UBE2D family members, and potential off-target effects

  • Quantitative proteomics should be employed to verify selective degradation

Using UBE2D4 as a PROTAC component:

  • UBE2D recruiters like EN67 could potentially be incorporated into PROTACs to target other proteins for degradation

  • Even partial engagement (~24%) of UBE2D in cells may be sufficient for PROTAC applications, as heterobifunctional compounds often demonstrate sub-stoichiometric activity

  • Both reactive and non-reactive PROTAC designs may be effective, as demonstrated with the NF500C and NF534 compounds

When developing and testing UBE2D4-related PROTACs, researchers should employ multiple complementary approaches to assess efficacy and specificity:

  • Western blotting to determine target protein levels

  • Proteomics to assess off-target effects

  • Dose-response and time-course experiments

  • Competition experiments with parental compounds

  • Control compounds with inactivated binding moieties

This field remains relatively unexplored for UBE2D4 specifically, but the principles established for other E2 enzymes provide a foundation for future development.

How should researchers interpret contradictory results between UBE2D4 studies in different model systems?

Contradictory results between UBE2D4 studies in different model systems require careful interpretation considering several factors:

Family member redundancy: The high sequence similarity between UBE2D1, UBE2D2, UBE2D3, and UBE2D4 means that the level of functional redundancy may vary between model systems. In systems where multiple UBE2D family members are expressed, knockdown of UBE2D4 alone may produce minimal phenotypes due to compensation by other family members. Comprehensive analysis should examine all UBE2D isoforms simultaneously or employ combined knockdown approaches.

Expression level variations: Different tissues and model systems may express UBE2D4 at varying levels relative to other UBE2D family members. RNA-seq analysis of expression patterns across tissues can help explain tissue-specific differences in experimental outcomes . Experimental designs should include quantification of baseline expression of all UBE2D family members in the models being compared.

E3 ligase partner availability: UBE2D4 functions in concert with E3 ligases, and the repertoire of available E3 partners differs between cell types and model organisms. This can lead to substantial differences in UBE2D4 function and substrate specificity between systems. Researchers should characterize the E3 landscape in their experimental systems when interpreting results.

Methodological differences: Variation in knockdown efficiency, expression levels of transgenes, timing of interventions, and analysis methods can all contribute to apparent contradictions. Detailed reporting of methodological parameters and standardization of approaches between research groups can help address these issues.

When faced with contradictory results, researchers should conduct carefully controlled experiments that directly compare different model systems using identical methodological approaches. Cross-validation using multiple independent techniques provides the strongest evidence for UBE2D4 functions.

What statistical approaches are most appropriate for analyzing UBE2D4 proteomics data?

Analysis of proteomics data related to UBE2D4 requires robust statistical approaches to identify true biological signals amid technical variation. Several considerations are important:

For differential abundance analysis:

  • Multiple testing correction is essential due to the large number of proteins typically identified in proteomics experiments

  • Commonly used approaches include Benjamini-Hochberg false discovery rate (FDR) correction

  • Thresholds for significance typically include both statistical significance (p-value < 0.01 or 0.05) and fold-change cutoffs

  • Volcano plots can effectively visualize proteins meeting both criteria

For pathway enrichment analysis:

  • Gene Ontology (GO) enrichment analysis helps identify biological processes affected by UBE2D4 modulation

  • Gene Set Enrichment Analysis (GSEA) can detect subtle but coordinated changes in functional groups of proteins

  • Protein-protein interaction network analysis helps identify clusters of functionally related proteins affected by UBE2D4

For integration with other data types:

  • Correlation analysis between proteomics and transcriptomics data can distinguish post-translational effects from transcriptional regulation

  • Linear modeling approaches can incorporate covariates (e.g., cell type, treatment duration) into the analysis

In UBE2D studies, proteomics analysis has revealed that knockdown of UBE2D/eff affects several categories of proteins, including proteasome components, chaperones, deubiquitinases, and ubiquitin ligases (upregulated), as well as peptidases, lipases, and secreted proteins (downregulated) . These findings were based on statistical analysis of TMT-based proteomics data comparing control and knockdown conditions, with appropriate corrections for multiple testing.

When designing the statistical analysis plan for UBE2D4 proteomics experiments, researchers should include proper replication (typically at least three biological replicates per condition), appropriate controls, and careful consideration of potential batch effects.

How can researchers distinguish between direct and indirect effects of UBE2D4 perturbation in experimental systems?

Distinguishing direct from indirect effects of UBE2D4 perturbation represents a significant challenge, particularly given its role in the ubiquitination pathway which affects numerous cellular processes. Several methodological approaches can help address this challenge:

Rescue experiments: Expression of RNAi-resistant UBE2D4 following knockdown can help identify which effects are specifically due to UBE2D4 loss. Effects that are reversed by UBE2D4 re-expression are more likely to be direct consequences of UBE2D4 activity. This approach has been used effectively in studies of UBE2D/eff knockdown in Drosophila .

Catalytically inactive mutants: Comparing effects of wild-type UBE2D4 with catalytically inactive mutants (e.g., active site cysteine mutants) can help distinguish between effects dependent on enzymatic activity versus scaffold functions.

Time-course experiments: Direct effects of UBE2D4 perturbation typically occur more rapidly than indirect consequences. Early time points after perturbation are more likely to reveal direct effects, while later time points may include numerous secondary adaptations.

Combined ubiquitinome and proteome analysis: Identifying proteins that show altered ubiquitination status shortly after UBE2D4 perturbation, before significant changes in total protein levels occur, can help identify direct ubiquitination targets.

In vitro validation: Reconstituted in vitro ubiquitination assays with purified components can definitively demonstrate direct ubiquitination of candidate substrates by UBE2D4 working with specific E3 ligases.

In the UBE2D literature, researchers have used TMT-based proteomics to identify proteins whose levels change upon UBE2D/eff knockdown and are subsequently rescued by human UBE2D2 expression . Proteins like Arc1, Arc2, Gnmt, and CG4594 showed this pattern and represent potential direct or closely linked targets of UBE2D-mediated regulation .

What are the most promising approaches for developing isoform-specific tools to study UBE2D4?

CRISPR-based tagging of endogenous UBE2D4: Inserting epitope tags or fluorescent proteins at the endogenous UBE2D4 locus allows specific tracking without overexpression artifacts. This approach requires careful design to target unique regions of the UBE2D4 gene and validation that the tag doesn't interfere with function.

Exploitation of non-conserved regions: While the catalytic domains of UBE2D family members are highly conserved, there may be subtle differences in surface residues or regulatory regions that could be exploited for selective targeting. Computational analysis and structural biology approaches can identify such regions.

Isoform-specific degraders: Development of selective UBE2D4 degraders (e.g., PROTACs) targeting any unique surface features could provide temporal control of UBE2D4 levels without affecting other family members. Covalent recruiters like those developed for the broader UBE2D family could potentially be refined for greater selectivity.

Single-cell analysis techniques: Methods that can analyze individual cells may reveal contexts where UBE2D4 is the predominant family member expressed, providing a natural system for studying its specific functions. Single-cell RNA-seq and proteomics approaches could identify such contexts.

Substrate-specific approaches: Identifying substrates preferentially ubiquitinated by UBE2D4 (perhaps due to specific E3 ligase partnerships) could provide indirect readouts of UBE2D4 activity. This requires comprehensive analysis of ubiquitination patterns following selective perturbation of each UBE2D family member.

The development of these tools will require collaborative efforts between structural biologists, chemists, geneticists, and cell biologists to overcome the challenge of distinguishing between highly similar protein family members.

What are the unexplored connections between UBE2D4 and age-related diseases?

The connection between UBE2D4 and age-related diseases represents a promising area for future research, with several intriguing directions:

Neurodegenerative disorders: Evidence already indicates that UBE2D family members influence protein aggregation in Huntington's disease models . Future research should examine potential roles in other proteinopathies like Alzheimer's, Parkinson's, and ALS. Gene expression analysis has revealed that proteome changes induced by UBE2D inhibition resemble profiles of cells experiencing proteotoxic stress, including those found in Parkinson's and Alzheimer's disease .

Sarcopenia and muscle aging: UBE2D knockdown in muscle reduces lifespan and proteasomal activity , suggesting potential involvement in age-related muscle decline. Investigating UBE2D4 expression and activity in aging human muscle could reveal connections to sarcopenia pathophysiology. The discovery that UBE2D/eff is necessary for maintaining proteasomal activity in skeletal muscle suggests it may play a protective role against age-related muscle protein quality decline.

Metabolic diseases: UBE2D enzymes regulate proteins involved in metabolism, including Gnmt (glycine N-methyltransferase) and enzymes involved in fatty acid beta-oxidation . This suggests potential connections to age-related metabolic disorders like diabetes and obesity that remain largely unexplored.

Cancer: Dysregulation of ubiquitination pathways is common in cancer, but specific roles of UBE2D4 are not well characterized. Investigation of cancer genomics databases for UBE2D4 alterations and correlation with patient outcomes could reveal unexpected connections.

Immunosenescence: The ubiquitin system plays important roles in immune function, and age-related decline in UBE2D4 activity could potentially contribute to immunosenescence, the age-related decline in immune function that increases susceptibility to infection and cancer.

Research approaches to explore these connections should include analysis of UBE2D4 expression in human tissues across the lifespan, genetic association studies in age-related diseases, animal models with tissue-specific modulation of UBE2D4, and mechanistic studies linking UBE2D4-mediated ubiquitination to specific disease processes.

Product Science Overview

Introduction to Ubiquitination

Ubiquitination is a critical post-translational modification process that occurs in all eukaryotic cells. It involves the covalent attachment of ubiquitin, a small 8.6 kDa protein, to specific residues on target proteins. This modification can occur as monoubiquitination, where a single ubiquitin molecule is attached, or polyubiquitination, where chains of ubiquitin molecules are formed. The primary function of polyubiquitination is to target proteins for degradation by the proteasome, although ubiquitination also plays significant roles in various cellular signaling pathways .

Role of Ubiquitin Conjugating Enzymes (E2)

The ubiquitination process is mediated by a cascade of three types of enzymes: E1 (ubiquitin-activating enzymes), E2 (ubiquitin-conjugating enzymes), and E3 (ubiquitin ligases). E2 enzymes are essential intermediates in this process, acting as ubiquitin donors for E3 ligases, which confer substrate specificity. The E2 enzymes are responsible for transferring ubiquitin from the E1 enzyme to the substrate protein, with the help of E3 ligases .

Ubiquitin Conjugating Enzyme E2D4 (UBE2D4)

UBE2D4, also known as Ubiquitin Conjugating Enzyme E2D4, is a member of the UBE2D family of E2 enzymes. This family includes UBE2D1, UBE2D2, UBE2D3, and UBE2D4, all of which share a high degree of sequence similarity and functional redundancy. UBE2D4 plays a crucial role in the ubiquitination of various target proteins, including receptor tyrosine kinases (RTKs) and components of the Hedgehog, TGFβ, and NFκB signaling pathways .

Functional Roles and Mechanisms

UBE2D4 is involved in several key cellular processes, including:

  • Transcriptional Control: UBE2D4 acts as a donor for the ubiquitination of histone tails by the Polycomb protein Ring1B and the DNA methylation regulator UHRF1. This activity is essential for the regulation of gene expression and chromatin structure .
  • DNA Repair: UBE2D4 plays a role in the DNA damage response by facilitating the ubiquitination of proteins involved in DNA repair pathways .
  • Tumor Suppression: UBE2D4 is involved in the regulation of the tumor suppressor protein p53, which is critical for maintaining cellular homeostasis and preventing cancer development .
Structural Insights

The structural details of UBE2D4 have been elucidated using homology modeling techniques. These studies have provided valuable insights into the enzyme’s active site and its interactions with ubiquitin and substrate proteins. Understanding the structure of UBE2D4 is essential for the development of novel therapeutic strategies targeting this enzyme for cancer treatment .

Recombinant UBE2D4

Recombinant UBE2D4 is produced using recombinant DNA technology, which involves the insertion of the UBE2D4 gene into an expression vector, followed by the expression of the protein in a suitable host organism, such as Escherichia coli. The recombinant protein is then purified using various chromatographic techniques to obtain a highly pure and active enzyme. Recombinant UBE2D4 is widely used in research to study the ubiquitination process and to develop potential therapeutic agents targeting this enzyme .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.